Skip to main content
Top
Published in: BMC Cancer 1/2018

Open Access 01-12-2018 | Research article

Novel role of O-glycosyltransferases GALNT3 and B3GNT3 in the self-renewal of pancreatic cancer stem cells

Authors: Srikanth Barkeer, Seema Chugh, Saswati Karmakar, Garima Kaushik, Sanchita Rauth, Satyanarayana Rachagani, Surinder K. Batra, Moorthy P. Ponnusamy

Published in: BMC Cancer | Issue 1/2018

Login to get access

Abstract

Background

Glycosylation plays a critical role in the aggressiveness of pancreatic cancer (PC). Emerging evidences indicate significant involvement of cancer stem cells (CSCs) in PC aggressiveness. However, the importance of glycosylation in pancreatic cancer stem cells (PCSCs) is yet to be addressed. Hence, we evaluated the potential role of glycosylation in maintenance of stemness of PCSCs.

Methods

Effect of glycosylation specific inhibitors on growth and PCSCs of PC cells was assessed by MTT assay and Side Population (SP) analysis. Isolated PCSCs/SP were characterized using molecular and functional assays. Expression of tumor-associated carbohydrate antigens (TACAs) was analyzed in PCSCs by western blotting. Effect of tunicamycin on PCSCs was analyzed by tumorsphere, clonogenicity, migration assay and immunoblotting for CSCs markers. The differential expression of glycogenes in PCSCs compared to non-CSCs were determined by RT-qPCR, immunoblotting and immunofluorescence. Co-expression of GALNT3 and B3GNT3 with CD44v6 was assessed in progression stages of KrasG12D; Pdx-1-Cre (KC) and KrasG12D; p53R172H; Pdx-1-Cre (KPC) tumors by immunofluorescence. Transient and CRISPR/Cas9 silencing of GALNT3 and B3GNT3 was performed to examine their effect on CSCs maintenance.

Results

Inhibition of glycosylation decreased growth and CSCs/SP in PC cells. PCSCs overexpressed CSC markers (CD44v6, ESA, SOX2, SOX9 and ABCG2), exhibited global expressional variation of TACAs and showed higher self-renewal potential. Specifically, N-glycosylation inhibition, significantly decreased tumorsphere formation, migration, and clonogenicity of PCSCs, as well as hypo-glycosylated CD44v6 and ESA. Of note, glycosyltransferases (GFs), GALNT3 and B3GNT3, were significantly overexpressed in PCSCs and co-expressed with CD44v6 at advanced PDAC stages in KC and KPC tumors. Further, GALNT3 and B3GNT3 knockdown led to a decrease in the expression of cell surface markers (CD44v6 and ESA) and self-renewal markers (SOX2 and OCT3/4) in PCSCs. Interestingly, CD44v6 was modified with sialyl Lewis a in PCSCs. Finally, CRISPR/Cas9-mediated GALNT3 KO significantly decreased self-renewal, clonogenicity, and migratory capacity in PCSCs.

Conclusions

Taken together, for the first time, our study showed the importance of glycosylation in mediating growth, stemness, and maintenance of PCSCs. These results indicate that elevated GALNT3 and B3GNT3 expression in PCSCs regulate stemness through modulating CSC markers.
Appendix
Available only for authorised users
Literature
1.
go back to reference Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74(11):2913–21.PubMedCrossRef Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74(11):2913–21.PubMedCrossRef
2.
go back to reference Jiang JH, Liu C, Cheng H, Lu Y, Qin Y, Xu YF, Xu J, Long J, Liu L, Ni QX, et al. Epithelial-mesenchymal transition in pancreatic cancer: is it a clinically significant factor? Biochim Biophys Acta. 2015;1855(1):43–9.PubMed Jiang JH, Liu C, Cheng H, Lu Y, Qin Y, Xu YF, Xu J, Long J, Liu L, Ni QX, et al. Epithelial-mesenchymal transition in pancreatic cancer: is it a clinically significant factor? Biochim Biophys Acta. 2015;1855(1):43–9.PubMed
3.
go back to reference Hezel AF, Kimmelman AC, Stanger BZ, Bardeesy N, Depinho RA. Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev. 2006;20(10):1218–49.PubMedCrossRef Hezel AF, Kimmelman AC, Stanger BZ, Bardeesy N, Depinho RA. Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev. 2006;20(10):1218–49.PubMedCrossRef
5.
go back to reference Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414(6859):105–11.PubMedCrossRef Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414(6859):105–11.PubMedCrossRef
6.
go back to reference Lobo NA, Shimono Y, Qian D, Clarke MF. The biology of cancer stem cells. Annu Rev Cell Dev Biol. 2007;23:675–99.PubMedCrossRef Lobo NA, Shimono Y, Qian D, Clarke MF. The biology of cancer stem cells. Annu Rev Cell Dev Biol. 2007;23:675–99.PubMedCrossRef
7.
go back to reference Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 2003;100(7):3983–8.PubMedPubMedCentralCrossRef Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 2003;100(7):3983–8.PubMedPubMedCentralCrossRef
8.
go back to reference Vaz AP, Ponnusamy MP, Batra SK. Cancer stem cells and therapeutic targets: an emerging field for cancer treatment. Drug Deliv Transl Res. 2013;3(2):113–20.PubMedPubMedCentralCrossRef Vaz AP, Ponnusamy MP, Batra SK. Cancer stem cells and therapeutic targets: an emerging field for cancer treatment. Drug Deliv Transl Res. 2013;3(2):113–20.PubMedPubMedCentralCrossRef
9.
go back to reference Mimeault M, Hauke R, Mehta PP, Batra SK. Recent advances in cancer stem/progenitor cell research: therapeutic implications for overcoming resistance to the most aggressive cancers. J Cell Mol Med. 2007;11(5):981–1011.PubMedPubMedCentralCrossRef Mimeault M, Hauke R, Mehta PP, Batra SK. Recent advances in cancer stem/progenitor cell research: therapeutic implications for overcoming resistance to the most aggressive cancers. J Cell Mol Med. 2007;11(5):981–1011.PubMedPubMedCentralCrossRef
10.
go back to reference Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, Guba M, Bruns CJ, Heeschen C. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell. 2007;1(3):313–23.PubMedCrossRef Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, Guba M, Bruns CJ, Heeschen C. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell. 2007;1(3):313–23.PubMedCrossRef
11.
go back to reference Zhan HX, Xu JW, Wu D, Zhang TP, Hu SY. Pancreatic cancer stem cells: new insight into a stubborn disease. Cancer Lett. 2015;357(2):429–37.PubMedCrossRef Zhan HX, Xu JW, Wu D, Zhang TP, Hu SY. Pancreatic cancer stem cells: new insight into a stubborn disease. Cancer Lett. 2015;357(2):429–37.PubMedCrossRef
12.
go back to reference Wang YC, Lin V, Loring JF, Peterson SE. The ‘sweet’ spot of cellular pluripotency: protein glycosylation in human pluripotent stem cells and its applications in regenerative medicine. Expert Opin Biol Ther. 2015;15(5):679–87.PubMedCrossRef Wang YC, Lin V, Loring JF, Peterson SE. The ‘sweet’ spot of cellular pluripotency: protein glycosylation in human pluripotent stem cells and its applications in regenerative medicine. Expert Opin Biol Ther. 2015;15(5):679–87.PubMedCrossRef
13.
go back to reference Jang H, Kim TW, Yoon S, Choi SY, Kang TW, Kim SY, Kwon YW, Cho EJ, Youn HD. O-GlcNAc regulates pluripotency and reprogramming by directly acting on core components of the pluripotency network. Cell Stem Cell. 2012;11(1):62–74.PubMedCrossRef Jang H, Kim TW, Yoon S, Choi SY, Kang TW, Kim SY, Kwon YW, Cho EJ, Youn HD. O-GlcNAc regulates pluripotency and reprogramming by directly acting on core components of the pluripotency network. Cell Stem Cell. 2012;11(1):62–74.PubMedCrossRef
14.
go back to reference Varki A, Lowe JB. Biological Roles of Glycans. In: Varki A, Cummings RD, Esko JD, Freeze HH, Stanley P, Bertozzi CR, Hart GW, Etzler ME, editors. Essentials of Glycobiology. 2nd ed. Cold Spring Harbor: Cold Spring Harbor Laboratory Press; 2009. Varki A, Lowe JB. Biological Roles of Glycans. In: Varki A, Cummings RD, Esko JD, Freeze HH, Stanley P, Bertozzi CR, Hart GW, Etzler ME, editors. Essentials of Glycobiology. 2nd ed. Cold Spring Harbor: Cold Spring Harbor Laboratory Press; 2009.
15.
go back to reference Pinho SS, Reis CA. Glycosylation in cancer: mechanisms and clinical implications. Nat Rev Cancer. 2015;15(9):540–55.PubMedCrossRef Pinho SS, Reis CA. Glycosylation in cancer: mechanisms and clinical implications. Nat Rev Cancer. 2015;15(9):540–55.PubMedCrossRef
16.
go back to reference Chugh S, Gnanapragassam VS, Jain M, Rachagani S, Ponnusamy MP, Batra SK. Pathobiological implications of mucin glycans in cancer: sweet poison and novel targets. Biochim Biophys Acta. 2015;1856(2):211–25.PubMedPubMedCentral Chugh S, Gnanapragassam VS, Jain M, Rachagani S, Ponnusamy MP, Batra SK. Pathobiological implications of mucin glycans in cancer: sweet poison and novel targets. Biochim Biophys Acta. 2015;1856(2):211–25.PubMedPubMedCentral
17.
go back to reference Chugh S, Barkeer S, Rachagani S, Nimmakayala RK, Perumal N, Pothuraju R, Atri P, Mahapatra S, Thapa I, Talmon GA, et al. Disruption of C1galt1 gene promotes development and metastasis of pancreatic adenocarcinomas in mice. Gastroenterology. 2018;155(5):1608–24.PubMedCrossRef Chugh S, Barkeer S, Rachagani S, Nimmakayala RK, Perumal N, Pothuraju R, Atri P, Mahapatra S, Thapa I, Talmon GA, et al. Disruption of C1galt1 gene promotes development and metastasis of pancreatic adenocarcinomas in mice. Gastroenterology. 2018;155(5):1608–24.PubMedCrossRef
18.
go back to reference Radhakrishnan P, Dabelsteen S, Madsen FB, Francavilla C, Kopp KL, Steentoft C, Vakhrushev SY, Olsen JV, Hansen L, Bennett EP, et al. Immature truncated O-glycophenotype of cancer directly induces oncogenic features. Proc Natl Acad Sci U S A. 2014;111(39):E4066–75.PubMedPubMedCentralCrossRef Radhakrishnan P, Dabelsteen S, Madsen FB, Francavilla C, Kopp KL, Steentoft C, Vakhrushev SY, Olsen JV, Hansen L, Bennett EP, et al. Immature truncated O-glycophenotype of cancer directly induces oncogenic features. Proc Natl Acad Sci U S A. 2014;111(39):E4066–75.PubMedPubMedCentralCrossRef
19.
go back to reference Radhakrishnan P, Grandgenett PM, Mohr AM, Bunt SK, Yu F, Chowdhury S, Hollingsworth MA. Expression of core 3 synthase in human pancreatic cancer cells suppresses tumor growth and metastasis. Int J Cancer. 2013;133(12):2824–33.PubMed Radhakrishnan P, Grandgenett PM, Mohr AM, Bunt SK, Yu F, Chowdhury S, Hollingsworth MA. Expression of core 3 synthase in human pancreatic cancer cells suppresses tumor growth and metastasis. Int J Cancer. 2013;133(12):2824–33.PubMed
20.
go back to reference Gautam SK, Kumar S, Cannon A, Hall B, Bhatia R, Nasser MW, Mahapatra S, Batra SK, Jain M. MUC4 mucin- a therapeutic target for pancreatic ductal adenocarcinoma. Expert Opin Ther Targets. 2017;21(7):657–69.PubMedPubMedCentralCrossRef Gautam SK, Kumar S, Cannon A, Hall B, Bhatia R, Nasser MW, Mahapatra S, Batra SK, Jain M. MUC4 mucin- a therapeutic target for pancreatic ductal adenocarcinoma. Expert Opin Ther Targets. 2017;21(7):657–69.PubMedPubMedCentralCrossRef
21.
23.
go back to reference Joshi S, Kumar S, Choudhury A, Ponnusamy MP, Batra SK. Altered mucins (MUC) trafficking in benign and malignant conditions. Oncotarget. 2014;5(17):7272–84.PubMedPubMedCentralCrossRef Joshi S, Kumar S, Choudhury A, Ponnusamy MP, Batra SK. Altered mucins (MUC) trafficking in benign and malignant conditions. Oncotarget. 2014;5(17):7272–84.PubMedPubMedCentralCrossRef
24.
go back to reference Kaur S, Kumar S, Momi N, Sasson AR, Batra SK. Mucins in pancreatic cancer and its microenvironment. Nat Rev Gastroenterol Hepatol. 2013;10(10):607–20.PubMedPubMedCentralCrossRef Kaur S, Kumar S, Momi N, Sasson AR, Batra SK. Mucins in pancreatic cancer and its microenvironment. Nat Rev Gastroenterol Hepatol. 2013;10(10):607–20.PubMedPubMedCentralCrossRef
25.
go back to reference Barkeer S, Chugh S, Batra SK, Ponnusamy MP. Glycosylation of Cancer stem cells: function in Stemness, tumorigenesis, and Metastasis. Neoplasia. 2018;20(8):813–25.PubMedPubMedCentralCrossRef Barkeer S, Chugh S, Batra SK, Ponnusamy MP. Glycosylation of Cancer stem cells: function in Stemness, tumorigenesis, and Metastasis. Neoplasia. 2018;20(8):813–25.PubMedPubMedCentralCrossRef
26.
go back to reference Terao N, Takamatsu S, Minehira T, Sobajima T, Nakayama K, Kamada Y, Miyoshi E. Fucosylation is a common glycosylation type in pancreatic cancer stem cell-like phenotypes. World J Gastroenterol. 2015;21(13):3876–87.PubMedPubMedCentralCrossRef Terao N, Takamatsu S, Minehira T, Sobajima T, Nakayama K, Kamada Y, Miyoshi E. Fucosylation is a common glycosylation type in pancreatic cancer stem cell-like phenotypes. World J Gastroenterol. 2015;21(13):3876–87.PubMedPubMedCentralCrossRef
27.
go back to reference Schultz MJ, Holdbrooks AT, Chakraborty A, Grizzle WE, Landen CN, Buchsbaum DJ, Conner MG, Arend RC, Yoon KJ, Klug CA, et al. The tumor-associated glycosyltransferase ST6Gal-I regulates stem cell transcription factors and confers a Cancer stem cell phenotype. Cancer Res. 2016;76(13):3978–88.PubMedPubMedCentralCrossRef Schultz MJ, Holdbrooks AT, Chakraborty A, Grizzle WE, Landen CN, Buchsbaum DJ, Conner MG, Arend RC, Yoon KJ, Klug CA, et al. The tumor-associated glycosyltransferase ST6Gal-I regulates stem cell transcription factors and confers a Cancer stem cell phenotype. Cancer Res. 2016;76(13):3978–88.PubMedPubMedCentralCrossRef
28.
go back to reference Li D, Su D, Xue L, Liu Y, Pang W. Establishment of pancreatic cancer stem cells by flow cytometry and their biological characteristics. Int J Clin Exp Pathol. 2015;8(9):11218–23.PubMedPubMedCentral Li D, Su D, Xue L, Liu Y, Pang W. Establishment of pancreatic cancer stem cells by flow cytometry and their biological characteristics. Int J Clin Exp Pathol. 2015;8(9):11218–23.PubMedPubMedCentral
29.
go back to reference Vaz AP, Ponnusamy MP, Rachagani S, Dey P, Ganti AK, Batra SK. Novel role of pancreatic differentiation 2 in facilitating self-renewal and drug resistance of pancreatic cancer stem cells. Br J Cancer. 2014;111(3):486–96.PubMedPubMedCentralCrossRef Vaz AP, Ponnusamy MP, Rachagani S, Dey P, Ganti AK, Batra SK. Novel role of pancreatic differentiation 2 in facilitating self-renewal and drug resistance of pancreatic cancer stem cells. Br J Cancer. 2014;111(3):486–96.PubMedPubMedCentralCrossRef
30.
go back to reference Lakshmanan I, Rachagani S, Hauke R, Krishn SR, Paknikar S, Seshacharyulu P, Karmakar S, Nimmakayala RK, Kaushik G, Johansson SL, et al. MUC5AC interactions with integrin beta4 enhances the migration of lung cancer cells through FAK signaling. Oncogene. 2016;35(31):4112–21.PubMedPubMedCentralCrossRef Lakshmanan I, Rachagani S, Hauke R, Krishn SR, Paknikar S, Seshacharyulu P, Karmakar S, Nimmakayala RK, Kaushik G, Johansson SL, et al. MUC5AC interactions with integrin beta4 enhances the migration of lung cancer cells through FAK signaling. Oncogene. 2016;35(31):4112–21.PubMedPubMedCentralCrossRef
31.
go back to reference Kumar S, Das S, Rachagani S, Kaur S, Joshi S, Johansson SL, Ponnusamy MP, Jain M, Batra SK. NCOA3-mediated upregulation of mucin expression via transcriptional and post-translational changes during the development of pancreatic cancer. Oncogene. 2015;34(37):4879–89.PubMedCrossRef Kumar S, Das S, Rachagani S, Kaur S, Joshi S, Johansson SL, Ponnusamy MP, Jain M, Batra SK. NCOA3-mediated upregulation of mucin expression via transcriptional and post-translational changes during the development of pancreatic cancer. Oncogene. 2015;34(37):4879–89.PubMedCrossRef
32.
go back to reference Rachagani S, Torres MP, Kumar S, Haridas D, Baine M, Macha MA, Kaur S, Ponnusamy MP, Dey P, Seshacharyulu P, et al. Mucin (Muc) expression during pancreatic cancer progression in spontaneous mouse model: potential implications for diagnosis and therapy. J Hematol Oncol. 2012;5:68.PubMedPubMedCentralCrossRef Rachagani S, Torres MP, Kumar S, Haridas D, Baine M, Macha MA, Kaur S, Ponnusamy MP, Dey P, Seshacharyulu P, et al. Mucin (Muc) expression during pancreatic cancer progression in spontaneous mouse model: potential implications for diagnosis and therapy. J Hematol Oncol. 2012;5:68.PubMedPubMedCentralCrossRef
33.
go back to reference Hirschhaeuser F, Menne H, Dittfeld C, West J, Mueller-Klieser W, Kunz-Schughart LA. Multicellular tumor spheroids: an underestimated tool is catching up again. J Biotechnol. 2010;148(1):3–15.PubMedCrossRef Hirschhaeuser F, Menne H, Dittfeld C, West J, Mueller-Klieser W, Kunz-Schughart LA. Multicellular tumor spheroids: an underestimated tool is catching up again. J Biotechnol. 2010;148(1):3–15.PubMedCrossRef
34.
go back to reference Mallard BW, Tiralongo J. Cancer stem cell marker glycosylation: nature, function and significance. Glycoconj J. 2017;34(4):441–52.PubMedCrossRef Mallard BW, Tiralongo J. Cancer stem cell marker glycosylation: nature, function and significance. Glycoconj J. 2017;34(4):441–52.PubMedCrossRef
35.
go back to reference Pauli C, Munz M, Kieu C, Mack B, Breinl P, Wollenberg B, Lang S, Zeidler R, Gires O. Tumor-specific glycosylation of the carcinoma-associated epithelial cell adhesion molecule EpCAM in head and neck carcinomas. Cancer Lett. 2003;193(1):25–32.PubMedCrossRef Pauli C, Munz M, Kieu C, Mack B, Breinl P, Wollenberg B, Lang S, Zeidler R, Gires O. Tumor-specific glycosylation of the carcinoma-associated epithelial cell adhesion molecule EpCAM in head and neck carcinomas. Cancer Lett. 2003;193(1):25–32.PubMedCrossRef
36.
go back to reference Fitzgerald TL, McCubrey JA. Pancreatic cancer stem cells: association with cell surface markers, prognosis, resistance, metastasis and treatment. Adv Biol Regul. 2014;56:45–50.PubMedCrossRef Fitzgerald TL, McCubrey JA. Pancreatic cancer stem cells: association with cell surface markers, prognosis, resistance, metastasis and treatment. Adv Biol Regul. 2014;56:45–50.PubMedCrossRef
37.
go back to reference Lei Y, Wang S, Ren B, Wang J, Chen J, Lu J, Zhan S, Fu Y, Huang L, Tan J. CHOP favors endoplasmic reticulum stress-induced apoptosis in hepatocellular carcinoma cells via inhibition of autophagy. PLoS One. 2017;12(8):e0183680.PubMedPubMedCentralCrossRef Lei Y, Wang S, Ren B, Wang J, Chen J, Lu J, Zhan S, Fu Y, Huang L, Tan J. CHOP favors endoplasmic reticulum stress-induced apoptosis in hepatocellular carcinoma cells via inhibition of autophagy. PLoS One. 2017;12(8):e0183680.PubMedPubMedCentralCrossRef
38.
go back to reference Schonthal AH. Endoplasmic reticulum stress: its role in disease and novel prospects for therapy. Scientifica (Cairo). 2012;2012:857516. Schonthal AH. Endoplasmic reticulum stress: its role in disease and novel prospects for therapy. Scientifica (Cairo). 2012;2012:857516.
39.
go back to reference Nami B, Donmez H, Kocak N. Tunicamycin-induced endoplasmic reticulum stress reduces in vitro subpopulation and invasion of CD44+/CD24- phenotype breast cancer stem cells. Exp Toxicol Pathol. 2016;68(7):419–26.PubMedCrossRef Nami B, Donmez H, Kocak N. Tunicamycin-induced endoplasmic reticulum stress reduces in vitro subpopulation and invasion of CD44+/CD24- phenotype breast cancer stem cells. Exp Toxicol Pathol. 2016;68(7):419–26.PubMedCrossRef
40.
go back to reference Bennett EP, Mandel U, Clausen H, Gerken TA, Fritz TA, Tabak LA. Control of mucin-type O-glycosylation: a classification of the polypeptide GalNAc-transferase gene family. Glycobiology. 2012;22(6):736–56.PubMedCrossRef Bennett EP, Mandel U, Clausen H, Gerken TA, Fritz TA, Tabak LA. Control of mucin-type O-glycosylation: a classification of the polypeptide GalNAc-transferase gene family. Glycobiology. 2012;22(6):736–56.PubMedCrossRef
41.
go back to reference He M, Wu C, Xu J, Guo H, Yang H, Zhang X, Sun J, Yu D, Zhou L, Peng T, et al. A genome wide association study of genetic loci that influence tumour biomarkers cancer antigen 19-9, carcinoembryonic antigen and alpha fetoprotein and their associations with cancer risk. Gut. 2014;63(1):143–51.PubMedCrossRef He M, Wu C, Xu J, Guo H, Yang H, Zhang X, Sun J, Yu D, Zhou L, Peng T, et al. A genome wide association study of genetic loci that influence tumour biomarkers cancer antigen 19-9, carcinoembryonic antigen and alpha fetoprotein and their associations with cancer risk. Gut. 2014;63(1):143–51.PubMedCrossRef
42.
go back to reference Yeh JC, Hiraoka N, Petryniak B, Nakayama J, Ellies LG, Rabuka D, Hindsgaul O, Marth JD, Lowe JB, Fukuda M. Novel sulfated lymphocyte homing receptors and their control by a Core1 extension beta 1,3-N-acetylglucosaminyltransferase. Cell. 2001;105(7):957–69.PubMedCrossRef Yeh JC, Hiraoka N, Petryniak B, Nakayama J, Ellies LG, Rabuka D, Hindsgaul O, Marth JD, Lowe JB, Fukuda M. Novel sulfated lymphocyte homing receptors and their control by a Core1 extension beta 1,3-N-acetylglucosaminyltransferase. Cell. 2001;105(7):957–69.PubMedCrossRef
43.
go back to reference Shiraishi N, Natsume A, Togayachi A, Endo T, Akashima T, Yamada Y, Imai N, Nakagawa S, Koizumi S, Sekine S, et al. Identification and characterization of three novel beta 1,3-N-acetylglucosaminyltransferases structurally related to the beta 1,3-galactosyltransferase family. J Biol Chem. 2001;276(5):3498–507.PubMedCrossRef Shiraishi N, Natsume A, Togayachi A, Endo T, Akashima T, Yamada Y, Imai N, Nakagawa S, Koizumi S, Sekine S, et al. Identification and characterization of three novel beta 1,3-N-acetylglucosaminyltransferases structurally related to the beta 1,3-galactosyltransferase family. J Biol Chem. 2001;276(5):3498–507.PubMedCrossRef
44.
go back to reference Taniuchi K, Cerny RL, Tanouchi A, Kohno K, Kotani N, Honke K, Saibara T, Hollingsworth MA. Overexpression of GalNAc-transferase GalNAc-T3 promotes pancreatic cancer cell growth. Oncogene. 2011;30(49):4843–54.PubMedPubMedCentralCrossRef Taniuchi K, Cerny RL, Tanouchi A, Kohno K, Kotani N, Honke K, Saibara T, Hollingsworth MA. Overexpression of GalNAc-transferase GalNAc-T3 promotes pancreatic cancer cell growth. Oncogene. 2011;30(49):4843–54.PubMedPubMedCentralCrossRef
45.
go back to reference Wang ZQ, Bachvarova M, Morin C, Plante M, Gregoire J, Renaud MC, Sebastianelli A, Bachvarov D. Role of the polypeptide N-acetylgalactosaminyltransferase 3 in ovarian cancer progression: possible implications in abnormal mucin O-glycosylation. Oncotarget. 2014;5(2):544–60.PubMedPubMedCentralCrossRef Wang ZQ, Bachvarova M, Morin C, Plante M, Gregoire J, Renaud MC, Sebastianelli A, Bachvarov D. Role of the polypeptide N-acetylgalactosaminyltransferase 3 in ovarian cancer progression: possible implications in abnormal mucin O-glycosylation. Oncotarget. 2014;5(2):544–60.PubMedPubMedCentralCrossRef
46.
go back to reference Chugh S, Meza J, Sheinin YM, Ponnusamy MP, Batra SK. Loss of N-acetylgalactosaminyltransferase 3 in poorly differentiated pancreatic cancer: augmented aggressiveness and aberrant ErbB family glycosylation. Br J Cancer. 2016;114(12):1376–86.PubMedPubMedCentralCrossRef Chugh S, Meza J, Sheinin YM, Ponnusamy MP, Batra SK. Loss of N-acetylgalactosaminyltransferase 3 in poorly differentiated pancreatic cancer: augmented aggressiveness and aberrant ErbB family glycosylation. Br J Cancer. 2016;114(12):1376–86.PubMedPubMedCentralCrossRef
47.
go back to reference Ponnusamy MP, Deb S, Dey P, Chakraborty S, Rachagani S, Senapati S, Batra SK. RNA polymerase II associated factor 1/PD2 maintains self-renewal by its interaction with Oct3/4 in mouse embryonic stem cells. Stem Cells. 2009;27(12):3001–11.PubMedPubMedCentral Ponnusamy MP, Deb S, Dey P, Chakraborty S, Rachagani S, Senapati S, Batra SK. RNA polymerase II associated factor 1/PD2 maintains self-renewal by its interaction with Oct3/4 in mouse embryonic stem cells. Stem Cells. 2009;27(12):3001–11.PubMedPubMedCentral
48.
go back to reference Dey P, Ponnusamy MP, Deb S, Batra SK. Human RNA polymerase II-association factor 1 (hPaf1/PD2) regulates histone methylation and chromatin remodeling in pancreatic cancer. PLoS One. 2011;6(10):e26926.PubMedPubMedCentralCrossRef Dey P, Ponnusamy MP, Deb S, Batra SK. Human RNA polymerase II-association factor 1 (hPaf1/PD2) regulates histone methylation and chromatin remodeling in pancreatic cancer. PLoS One. 2011;6(10):e26926.PubMedPubMedCentralCrossRef
49.
go back to reference Dey P, Rachagani S, Vaz AP, Ponnusamy MP, Batra SK. PD2/Paf1 depletion in pancreatic acinar cells promotes acinar-to-ductal metaplasia. Oncotarget. 2014;5(12):4480–91.PubMedPubMedCentralCrossRef Dey P, Rachagani S, Vaz AP, Ponnusamy MP, Batra SK. PD2/Paf1 depletion in pancreatic acinar cells promotes acinar-to-ductal metaplasia. Oncotarget. 2014;5(12):4480–91.PubMedPubMedCentralCrossRef
50.
go back to reference Vaz AP, Deb S, Rachagani S, Dey P, Muniyan S, Lakshmanan I, Karmakar S, Smith L, Johansson S, Lele S, et al. Overexpression of PD2 leads to increased tumorigenicity and metastasis in pancreatic ductal adenocarcinoma. Oncotarget. 2016;7(3):3317–31.PubMedCrossRef Vaz AP, Deb S, Rachagani S, Dey P, Muniyan S, Lakshmanan I, Karmakar S, Smith L, Johansson S, Lele S, et al. Overexpression of PD2 leads to increased tumorigenicity and metastasis in pancreatic ductal adenocarcinoma. Oncotarget. 2016;7(3):3317–31.PubMedCrossRef
51.
go back to reference Karmakar S, Seshacharyulu P, Lakshmanan I, Vaz AP, Chugh S, Sheinin YM, Mahapatra S, Batra SK, Ponnusamy MP. hPaf1/PD2 interacts with OCT3/4 to promote self-renewal of ovarian cancer stem cells. Oncotarget. 2017;8(9):14806–20.PubMedPubMedCentralCrossRef Karmakar S, Seshacharyulu P, Lakshmanan I, Vaz AP, Chugh S, Sheinin YM, Mahapatra S, Batra SK, Ponnusamy MP. hPaf1/PD2 interacts with OCT3/4 to promote self-renewal of ovarian cancer stem cells. Oncotarget. 2017;8(9):14806–20.PubMedPubMedCentralCrossRef
52.
go back to reference Zhang W, Hou T, Niu C, Song L, Zhang Y. B3GNT3 expression is a novel marker correlated with pelvic lymph node metastasis and poor clinical outcome in early-stage cervical Cancer. PLoS One. 2015;10(12):e0144360.PubMedPubMedCentralCrossRef Zhang W, Hou T, Niu C, Song L, Zhang Y. B3GNT3 expression is a novel marker correlated with pelvic lymph node metastasis and poor clinical outcome in early-stage cervical Cancer. PLoS One. 2015;10(12):e0144360.PubMedPubMedCentralCrossRef
53.
go back to reference Cerhan JR, Ansell SM, Fredericksen ZS, Kay NE, Liebow M, Call TG, Dogan A, Cunningham JM, Wang AH, Liu-Mares W, et al. Genetic variation in 1253 immune and inflammation genes and risk of non-Hodgkin lymphoma. Blood. 2007;110(13):4455–63.PubMedPubMedCentralCrossRef Cerhan JR, Ansell SM, Fredericksen ZS, Kay NE, Liebow M, Call TG, Dogan A, Cunningham JM, Wang AH, Liu-Mares W, et al. Genetic variation in 1253 immune and inflammation genes and risk of non-Hodgkin lymphoma. Blood. 2007;110(13):4455–63.PubMedPubMedCentralCrossRef
54.
go back to reference Ho WL, Che MI, Chou CH, Chang HH, Jeng YM, Hsu WM, Lin KH, Huang MC. B3GNT3 expression suppresses cell migration and invasion and predicts favorable outcomes in neuroblastoma. Cancer Sci. 2013;104(12):1600–8.PubMedCrossRefPubMedCentral Ho WL, Che MI, Chou CH, Chang HH, Jeng YM, Hsu WM, Lin KH, Huang MC. B3GNT3 expression suppresses cell migration and invasion and predicts favorable outcomes in neuroblastoma. Cancer Sci. 2013;104(12):1600–8.PubMedCrossRefPubMedCentral
55.
go back to reference Li C, Du Y, Yang Z, He L, Wang Y, Hao L, Ding M, Yan R, Wang J, Fan Z. GALNT1-mediated glycosylation and activation of sonic hedgehog signaling maintains the self-renewal and tumor-initiating capacity of bladder Cancer stem cells. Cancer Res. 2016;76(5):1273–83.PubMedCrossRef Li C, Du Y, Yang Z, He L, Wang Y, Hao L, Ding M, Yan R, Wang J, Fan Z. GALNT1-mediated glycosylation and activation of sonic hedgehog signaling maintains the self-renewal and tumor-initiating capacity of bladder Cancer stem cells. Cancer Res. 2016;76(5):1273–83.PubMedCrossRef
56.
go back to reference Guo H, Nagy T, Pierce M. Post-translational glycoprotein modifications regulate colon cancer stem cells and colon adenoma progression in Apc(min/+) mice through altered Wnt receptor signaling. J Biol Chem. 2014;289(45):31534–49.PubMedPubMedCentralCrossRef Guo H, Nagy T, Pierce M. Post-translational glycoprotein modifications regulate colon cancer stem cells and colon adenoma progression in Apc(min/+) mice through altered Wnt receptor signaling. J Biol Chem. 2014;289(45):31534–49.PubMedPubMedCentralCrossRef
57.
go back to reference Che MI, Huang J, Hung JS, Lin YC, Huang MJ, Lai HS, Hsu WM, Liang JT, Huang MC. beta1, 4-N-acetylgalactosaminyltransferase III modulates cancer stemness through EGFR signaling pathway in colon cancer cells. Oncotarget. 2014;5(11):3673–84.PubMedPubMedCentralCrossRef Che MI, Huang J, Hung JS, Lin YC, Huang MJ, Lai HS, Hsu WM, Liang JT, Huang MC. beta1, 4-N-acetylgalactosaminyltransferase III modulates cancer stemness through EGFR signaling pathway in colon cancer cells. Oncotarget. 2014;5(11):3673–84.PubMedPubMedCentralCrossRef
58.
go back to reference Matzke-Ogi A, Jannasch K, Shatirishvili M, Fuchs B, Chiblak S, Morton J, Tawk B, Lindner T, Sansom O, Alves F, et al. Inhibition of tumor growth and metastasis in pancreatic Cancer models by interference with CD44v6 signaling. Gastroenterology. 2016;150(2):513–25 e510.PubMedCrossRef Matzke-Ogi A, Jannasch K, Shatirishvili M, Fuchs B, Chiblak S, Morton J, Tawk B, Lindner T, Sansom O, Alves F, et al. Inhibition of tumor growth and metastasis in pancreatic Cancer models by interference with CD44v6 signaling. Gastroenterology. 2016;150(2):513–25 e510.PubMedCrossRef
59.
go back to reference Eom BW, Joo J, Park B, Jo MJ, Choi SH, Cho SJ, Ryu KW, Kim YW, Kook MC. Nomogram Incorporating CD44v6 and Clinicopathological Factors to Predict Lymph Node Metastasis for Early Gastric Cancer. PLoS One. 2016;11(8):e0159424.PubMedPubMedCentralCrossRef Eom BW, Joo J, Park B, Jo MJ, Choi SH, Cho SJ, Ryu KW, Kim YW, Kook MC. Nomogram Incorporating CD44v6 and Clinicopathological Factors to Predict Lymph Node Metastasis for Early Gastric Cancer. PLoS One. 2016;11(8):e0159424.PubMedPubMedCentralCrossRef
60.
go back to reference Hu B, Luo W, Hu RT, Zhou Y, Qin SY, Jiang HX. Meta-analysis of prognostic and clinical significance of CD44v6 in esophageal Cancer. Medicine (Baltimore). 2015;94(31):e1238.CrossRef Hu B, Luo W, Hu RT, Zhou Y, Qin SY, Jiang HX. Meta-analysis of prognostic and clinical significance of CD44v6 in esophageal Cancer. Medicine (Baltimore). 2015;94(31):e1238.CrossRef
61.
go back to reference Todaro M, Gaggianesi M, Catalano V, Benfante A, Iovino F, Biffoni M, Apuzzo T, Sperduti I, Volpe S, Cocorullo G, et al. CD44v6 is a marker of constitutive and reprogrammed cancer stem cells driving colon cancer metastasis. Cell Stem Cell. 2014;14(3):342–56.PubMedCrossRef Todaro M, Gaggianesi M, Catalano V, Benfante A, Iovino F, Biffoni M, Apuzzo T, Sperduti I, Volpe S, Cocorullo G, et al. CD44v6 is a marker of constitutive and reprogrammed cancer stem cells driving colon cancer metastasis. Cell Stem Cell. 2014;14(3):342–56.PubMedCrossRef
62.
go back to reference Ni J, Cozzi PJ, Hao JL, Beretov J, Chang L, Duan W, Shigdar S, Delprado WJ, Graham PH, Bucci J, et al. CD44 variant 6 is associated with prostate cancer metastasis and chemo−/radioresistance. Prostate. 2014;74(6):602–17.PubMedCrossRef Ni J, Cozzi PJ, Hao JL, Beretov J, Chang L, Duan W, Shigdar S, Delprado WJ, Graham PH, Bucci J, et al. CD44 variant 6 is associated with prostate cancer metastasis and chemo−/radioresistance. Prostate. 2014;74(6):602–17.PubMedCrossRef
63.
go back to reference Brazil JC, Liu R, Sumagin R, Kolegraff KN, Nusrat A, Cummings RD, Parkos CA, Louis NA. alpha3/4 Fucosyltransferase 3-dependent synthesis of Sialyl Lewis A on CD44 variant containing exon 6 mediates polymorphonuclear leukocyte detachment from intestinal epithelium during transepithelial migration. J Immunol. 2013;191(9):4804–17.PubMedCrossRef Brazil JC, Liu R, Sumagin R, Kolegraff KN, Nusrat A, Cummings RD, Parkos CA, Louis NA. alpha3/4 Fucosyltransferase 3-dependent synthesis of Sialyl Lewis A on CD44 variant containing exon 6 mediates polymorphonuclear leukocyte detachment from intestinal epithelium during transepithelial migration. J Immunol. 2013;191(9):4804–17.PubMedCrossRef
64.
go back to reference Hallouin F, Goupille C, Bureau V, Meflah K, Le Pendu J. Increased tumorigenicity of rat colon carcinoma cells after alpha1,2-fucosyltransferase FTA anti-sense cDNA transfection. Int J Cancer. 1999;80(4):606–11.PubMedCrossRef Hallouin F, Goupille C, Bureau V, Meflah K, Le Pendu J. Increased tumorigenicity of rat colon carcinoma cells after alpha1,2-fucosyltransferase FTA anti-sense cDNA transfection. Int J Cancer. 1999;80(4):606–11.PubMedCrossRef
65.
go back to reference Goupille C, Hallouin F, Meflah K, Le Pendu J. Increase of rat colon carcinoma cells tumorigenicity by alpha(1-2) fucosyltransferase gene transfection. Glycobiology. 1997;7(2):221–9.PubMedCrossRef Goupille C, Hallouin F, Meflah K, Le Pendu J. Increase of rat colon carcinoma cells tumorigenicity by alpha(1-2) fucosyltransferase gene transfection. Glycobiology. 1997;7(2):221–9.PubMedCrossRef
66.
go back to reference Singh R, Campbell BJ, Yu LG, Fernig DG, Milton JD, Goodlad RA, FitzGerald AJ, Rhodes JM. Cell surface-expressed Thomsen-Friedenreich antigen in colon cancer is predominantly carried on high molecular weight splice variants of CD44. Glycobiology. 2001;11(7):587–92.PubMedCrossRef Singh R, Campbell BJ, Yu LG, Fernig DG, Milton JD, Goodlad RA, FitzGerald AJ, Rhodes JM. Cell surface-expressed Thomsen-Friedenreich antigen in colon cancer is predominantly carried on high molecular weight splice variants of CD44. Glycobiology. 2001;11(7):587–92.PubMedCrossRef
Metadata
Title
Novel role of O-glycosyltransferases GALNT3 and B3GNT3 in the self-renewal of pancreatic cancer stem cells
Authors
Srikanth Barkeer
Seema Chugh
Saswati Karmakar
Garima Kaushik
Sanchita Rauth
Satyanarayana Rachagani
Surinder K. Batra
Moorthy P. Ponnusamy
Publication date
01-12-2018
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2018
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-018-5074-2

Other articles of this Issue 1/2018

BMC Cancer 1/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine